Anda di halaman 1dari 11

Premature aging and immune senescence

in HIV-infected children
Ketty Gianesina, Antoni Noguera-Julianb, Marisa Zanchettac,
Paola Del Biancoc, Maria Raffaella Petraraa, Riccardo Fregujaa,
Osvalda Rampond, Clàudia Fortunyb, Mireia Camóse, Elena Mozzod,
Carlo Giaquintod and Anita De Rossia,c

Objective: Several pieces of evidence indicate that HIV-infected adults undergo


premature aging. The effect of HIV and antiretroviral therapy (ART) exposure on the
aging process of HIV-infected children may be more deleterious since their immune
system coevolves from birth with HIV.
Design: Seventy-one HIV-infected (HIVþ), 65 HIV-exposed-uninfected (HEU), and 56
HIV-unexposed-uninfected (HUU) children, all aged 0–5 years, were studied for
biological aging and immune senescence.
Methods: Telomere length and T-cell receptor rearrangement excision circle levels
were quantified in peripheral blood cells by real-time PCR. CD4þ and CD8þ cells were
analysed for differentiation, senescence, and activation/exhaustion markers by flow
cytometry.
Results: Telomere lengths were significantly shorter in HIVþ than in HEU and HUU
children (overall, P < 0.001 adjusted for age); HIVþ ART-naive (42%) children had
shorter telomere length compared with children on ART (P ¼ 0.003 adjusted for age).
T-cell receptor rearrangement excision circle levels and CD8þ recent thymic emigrant
cells (CD45RAþCD31þ) were significantly lower in the HIVþ than in control groups
(overall, P ¼ 0.025 and P ¼ 0.005, respectively). Percentages of senescent
(CD28CD57þ), activated (CD38þHLA-DRþ), and exhausted (PD1þ) CD8þ cells were
significantly higher in HIVþ than in HEU and HUU children (P ¼ 0.004, P < 0.001, and
P < 0.001, respectively). Within the CD4þ cell subset, the percentage of senescent cells
did not differ between HIVþ and controls, but programmed cell death receptor-1
expression was upregulated in the former.
Conclusions: HIV-infected children exhibit premature biological aging with acceler-
ated immune senescence, which particularly affects the CD8þ cell subset. HIV infection
per se seems to influence the aging process, rather than exposure to ART for prophylaxis
or treatment. Copyright ß 2016 Wolters Kluwer Health, Inc. All rights reserved.

AIDS 2016, 30:1363–1373

Keywords: immune activation, immune senescence, microbial translocation,


pediatric HIV/AIDS, premature aging, telomere length, T-cell receptor
rearrangement excision circle

a
Department of Surgery, Oncology and Gastroenterology – DiSCOG, Section of Oncology and Immunology, Unit of Viral
Oncology and AIDS Reference Center, University of Padova, Padova, Italy, bUnitat d’Infectologia, Servei de Pediatria; Hospital
Sant Joan de Déu, Universitat de Barcelona, Barcelona, Spain, cIstituto Oncologico Veneto (IOV) – IRCCS, dDepartment of Mother
and Child Health, University of Padova, Padova, Italy, and eClinical Laboratory Department, Hematology Unit, Hospital San Joan
de Déu, Universitat de Barcelona, Barcelona, Spain.
Correspondence to Professor Anita De Rossi, Section of Oncology and Immunology, Unit of Viral Oncology and AIDS Reference
Center, Department of Surgery, Oncology and Gastroenterology – DiSCOG, University of Padova, Via Gattamelata 64, 35128
Padova, Italy.
Tel: +39 049 8215894; fax: +39 049 8072854; e-mail: anita.derossi@unipd.it
Received: 6 October 2015; revised: 25 February 2016; accepted: 7 March 2016.
DOI:10.1097/QAD.0000000000001093
ISSN 0269-9370 Copyright Q 2016 Wolters Kluwer Health, Inc. All rights reserved. This is an open-access article distributed under the terms
of the Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 License, where it is permissible to download and share the work
provided it is properly cited. The work cannot be changed in any way or used commercially. 1363
1364 AIDS 2016, Vol 30 No 9

Introduction comprehensive assessment of telomere shortening, a key


molecular marker of biological aging, together with the
The introduction of antiretroviral therapy (ART) has activation/senescent profile of T lymphocytes. In this
changed the natural history of pediatric HIV infection; study, we analysed biological aging in relation to immune
ART-based prophylaxis regimens have in fact reduced activation and senescence markers in a cohort of
mother-to-child transmission of HIV from 15–20% to perinatally HIV-infected children.
under 2% in high-income countries, and have also given
rise to substantial improvements in terms of survival and
quality of life in HIV-infected children [1,2]. HIV
infection is, therefore, now considered a chronic disease Methods
which persists for many decades [3]. However, despite
improvements in immune function and reduction of Ethic statement
AIDS-related complications, including opportunistic The study was approved by the Ethics Committees of the
infections and AIDS-associated malignancies, ART does Azienda Ospedaliera Padova (Prot. n.#2921P) and the
not restore full health. Many studies have demonstrated Hospital Sant Joan de Déu, Universitat de Barcelona
that ART-treated HIV-infected adults have a higher risk (Prot. n.#04–15); written informed consent was
of non-AIDS-related overall morbidity and mortality obtained for all children from their parents/guardians.
compared with age-matched HIV-uninfected individuals.
This increased risk is mainly because of a range of non- Study population
AIDS-defining illnesses associated with aging, including A total of 71 perinatally HIV-infected (HIVþ), 65 HIV-
malignancies [4–8], and it has been advanced that the exposed-uninfected (HEU), and 56 HIV-unexposed-
increase in non-AIDS-defining diseases among HIV- uninfected (HUU) children, all aged 0–5 years, were
infected patients may be because of premature aging [9]. included in this study. HIVþ and HEU children attended
The pathogenic mechanism underlying this increased risk the Department of Mother and Child Health, University
is still poorly understood. Chronic immune activation of Padova or the Infectious Diseases Unit, Pediatrics
because of the persistence of circulating HIV virions may Department, Hospital Sant Joan de Déu, Universitat de
play a key role in the senescent pathway. Activated cells Barcelona. For each HIVþ and HEU child, the first
undergo clonal expansion in response to viral persistence, cryopreserved sample available after birth was chosen for
resulting in differentiation and accumulation of non- the study. None of the HIVþ or HEU children was
functional senescent cells [10]. It has been also advanced breastfed. HUU children were recruited at Pediatric
that premature and accelerated aging in HIV-infected Emergency Department of Azienda Ospedaliera Padova
patients can be because of adverse effects of antiretroviral or Hospital Sant Joan de Déu, Universitat de Barcelona.
drugs. Nucleoside reverse transcriptase inhibitors have Exclusion criteria were malignancies, chronic infections,
been shown to inhibit telomerase activity in replicating sarcoidosis, diabetes mellitus type-1, rheumatoid arthritis,
cell lines in vitro, leading to accelerated shortening of and systemic lupus erythematosus.
telomere length [11,12].
Sample preparation
The clinical complications of HIV infection and ART Peripheral blood mononuclear cells (PBMC) were
treatment in children may be more serious than in adults. isolated from ethylenediaminetetraacetic acid-treated
The course of vertically transmitted infection in infants is peripheral blood (2–5 ml) by centrifugation on a
characterized by faster disease progression and shorter Ficoll-Paque gradient (Pharmacia, Uppsala, Sweden).
time to AIDS, compared with adults. After infection, PBMC were cryopreserved and plasma samples were
plasma HIV-RNA levels are higher in infants than in stored in liquid nitrogen and at 808C, until use.
adults. In addition, they persist at high levels and decline
slowly with age in the absence of ART [13,14], whereas Telomere length measurement by quantitative
in adults control of viral load is reached a few weeks after real-time PCR
infection [15]. This slower control of viral replication is Relative telomere length was determined by mono-
probably because of the fact that the immune system is still chrome quantitative multiplex PCR assay [24] with
maturing. The innate immunity in children is of minor modifications. Each PCR reaction was performed
particular importance and plays a critical role in HIV in a final volume of 25 ml containing 5 ml sample
pathogenesis [16–18], as the adaptive immune system is (2 ng DNA/ml) and 20 ml reaction mix containing
still developing [19]. HIV infection since birth together 0.75  SYBR GreenI (Invitrogen, Italy), 10 mmol/l
with long-term exposure to ART may affect premature Tris-hydrochloric acid pH 8.3, 50 mmol/l potassium
aging and immune senescence in HIV-infected children chloride, 3 mmol/l magnesium chloride, 0.2 mmol/l
even more than in adults. each deoxynucleotide (dNTP) (Applied Biosystems,
Foster City, California, USA), 1 mmol/l dithiothreitol,
To date, few data are available on premature aging in 0.625 U AmpliTaq Gold DNA polymerase, 1% dimethyl
HIV-infected children [20–23] and no reports give a sulfoxide (Sigma-Aldrich, St Louis, Missouri, USA), and
Pediatric HIV infection and aging Gianesin et al. 1365

900 nmol/l of each of the primers. Telomere and D Related (HLA-DR)-APC, and CD28-APC (Miltenyi
albumin gene primers sequences are described in [24]. Biotec, Auburn, California, USA). Appropriate isotypic
PCR reactions were performed on a LightCycler480 controls (mouse IgG1-PE, IgG2b-APC, and IgG1k-
real-time PCR detection system (Roche Applied PECy7) were used to evaluate nonspecific staining. Cells
Science, Mannheim, Germany). The thermal cycling were then washed and resuspended in phosphate-
profile was 15 min at 958C, two cycles of 15 s at 948C, 15 s buffered saline supplemented with 1% paraformalde-
at 498C, followed by 40 cycles of 15 s at 948C, 10 s hyde. All samples were analysed using LSRII Flow
at 628C, 15 s at 748C, 10 s at 848C, 15 s at 888C, with cytometer (Becton-Dickinson). A total of 100 000 events
signal acquisition at the end of both the 748C and 888C were collected in the lymphocyte gate using morpho-
steps. A standard curve was generated at each PCR run, logical parameters (forward and side-scatter). Data were
consisting of DNA from the RAJI cell line, serially processed with FACSDivaSoftware (Becton-Dickinson)
diluted from 100 to 0.41 ng/ml [25]. LightCycler raw and analysed using KaluzaAnalyzing Software v.1.2
text files were converted using the LC480Conversion (Beckman Coulter) (Supplementary Figure 1, http://
free software (http://www.hartfaalcentrum.nl/index. links.lww.com/QAD/A903). Samples for flow-cytome-
php?main¼files&fileName¼ LC480Conversion.zip&de try analysis were available for 24 HIVþ children, 21
scription¼LC480%20Conversion&sub¼LC480Convers HEU, and 18 HUU children. The characteristics of
ion), and the data were analysed using LinRegPCR free these subgroups are given in Supplementary Table 1,
software [26]. All samples were blindly and consecutively http://links.lww.com/QAD/A904.
run in triplicate together with reference samples. The
intra and interassay reproducibility of both telomere and Quantification of soluble markers
albumin PCR results was evaluated using dilutions of the Plasma levels of soluble CD14 (sCD14), IL-6, and TNFa
reference curve. Telomere length values were calculated were quantified with commercially available assays
as telomere/single-copy gene (T/S) ratio, as previously (Human sCD14, IL-6, and TNFa Quantikine ELI-SA;
described [25]. The intra and interassay variability of R&D Systems, Minneapolis, Minnesota, USA) according
T/S values was evaluated using reference samples; to the manufacturer’s protocol. Samples for analysis of
coefficients of variation were 3.98% or less and 8.14% soluble markers were available for 24 HIVþ, 21 HEU, and
or less, respectively. 18 HUU children.

T-cell receptor rearrangement excision circle Statistical analysis


levels quantification Unadjusted comparisons of continuous variable distri-
Thymic output in PBMC was studied by measurement of butions among groups were assessed with the Kruskal–
T-cell receptor rearrangement excision circle (TREC) Wallis nonparametric test, and the associations
levels by real-time PCR, as previously described [27]. between categorical variables were analysed by the
TREC levels were expressed as the number of TREC x2 test. Spearman’s r coefficient (rs) was used for
copies/105 PBMC. correlations. Normal distributions for telomere
length and TREC levels were visually checked by
Viral load quantification quantile–quantile plots. Linear regression models esti-
Plasma HIV-RNA levels were determined in all HIV- mated the telomere length and TREC levels in HIVþ,
infected children using the COBAS Taqman HIV-1 test HEU, and HUU children, ART exposure and naive
(Roche, Branchburg, New Jersey, USA). The lower limit groups, log-transformed HIV-RNA and HIV-DNA
of detection was 50 HIV-RNA copies/ml. HIV-DNA covariates, adjusted for age and its interaction with
levels in PBMC were measured by real-time PCR, and groups. Samples with undetectable plasma HIV-RNA
expressed as HIV-DNA copies/106 PBMC as previously were assigned a value of 20 copies/ml to include them in
described [27]. the statistical analyses. All statistical analyses were
performed with Statistical Analysis Software (SAS)
Flow cytometry analysis (Release 9.2; SAS Institute, Cary, North Carolina,
T-cell phenotyping was performed on cryopreserved USA). Adjustments for multiple testing with Hochberg’s
PBMC. Cells were thawed, washed, stained for 20 min in correction were made for comparisons of CD4þ and
the dark with the Live/Dead Fixable Near-IR Dead CD8þ cell subsets among groups. All P values were two-
Cell Stain Kit (Life Technologies, Carlsbad, California, tailed, and were considered significant at less than 0.05.
USA) and with fluorescent-conjugated mononuclear
antibodies CD3-fluorescein isothiocyanate, CD31-
phycoerythrin (PE), CD38-PE, CD57-PE, CD45RA-
allophycocyanin (APC) and programmed cell death Results
receptor (PD)-1-PECy7 (Becton-Dickinson, San Diego,
California, USA), CD27-PECy7 (Beckman Coulter, Characteristics of study population
Fullerton, California, USA) and CD4-VioBlue, The characteristics of the study population are listed in
CD8-VioGreen, Human Leukocyte Antigen - antigen Table 1. The median age of HIVþ children was 3.11
1366 AIDS 2016, Vol 30 No 9

[interquartile range (IQR) 1.41–4.48], 1.73 (0.99–3.20) but not with HIV-DNA levels (b ¼ 0.0004, P ¼ 0.490),
for HEU, and 1.85 (0.85–3.46) years for HUU children. and were significantly lower in ART-naive children
Thirty of the 71 (42%) HIVþ children were ART-naive, (b ¼ 0.3913, P ¼ 0.003). In the final multivariate
the others were on ART [median time 18 (11–36.5) model, including all variables, the stepwise method
months]. ART-naive HIVþ children were younger than selected only ART exposure as a significant predictor
those on therapy [1.70 (0.69–3.59) and 3.62 (2.17–4.81) variable of higher mean telomere length value
years, respectively; P ¼ 0.004]. Sixty-one of the 65 (P ¼ 0.039).
(93.8%) HEU children had been exposed to ART
prophylaxis during gestation [median 34 (17–38) weeks] Thymic output is lower in HIV-infected children
and received postnatal prophylaxis [6 weeks of zidovudine than in controls
(ZDV) monotherapy]. Of the 61 ART-treated pregnant HEU and HUU children had higher TREC levels than
women, 40 (65.6%) had received triple therapy based on HIVþ children [5409 (3470–6600), 5370 (2380–8102),
ZDV þ lamivudine (3TC) þ nevirapine (NVP), 17 3498 (2051–6780)] TREC copies/105 PBMC, respec-
(27.8%) ZDV þ 3TC þ protease inhibitor, and the tively [P ¼ 0.018 not age adjusted (Fig. 1e); P ¼ 0.025
remaining 4 (6.6%) a regimen based on tenofovir age-adjusted]. TREC levels decreased significantly with
(TDF) þ emtricitabine (FTC) þ protease inhibitor. increasing age in HEU and HUU groups (b ¼ 61,
P ¼ 0.009 and b ¼ 86, P ¼ 0.001, respectively), but not
Telomere length is shorter in HIV-infected in HIVþ children (b ¼ 17, P ¼ 0.353) (Fig. 1f). No
children than in controls significant differences in TREC dynamics were found
The median telomere length value in PBMC was between ART-treated and ART-naive children (Fig. 1g
significantly lower in HIVþ than in HEU and HUU and h).
children, being 2.21 (1.94–2.58), 2.63 (2.25–3.21), and
2.88 (2.49–3.1), respectively [P < 0.001 not age adjusted Phenotypic T-cell alterations occur early in HIV-
(Fig. 1a); P < 0.001 age adjusted]. Telomere length values infected children
significantly decreased with age in HEU and HUU There were no differences in the frequencies of CD3þ
(regression coefficient (b) ¼ 0.0102, P ¼ 0.008 and cells among the three groups (P ¼ 0.590) (Table 2). The
b ¼ 0.0100, P ¼ 0.011, respectively), but not in HIVþ percentages of total CD4þ cells were lower in the HIVþ
children (b ¼ 0.0018, P ¼ 0.587) (Fig. 1b). Of note, in than in the control groups (P ¼ 0.001). Within CD4þ
the HIVþ group, ART-naive children had shorter cells, HIVþand control groups did not significantly differ
telomere length compared with those on ART [2.11 in percentages of naive (CD45RAþCD27þ), central
(1.75–2.37) ‘vs.’ 2.46 (2.07–2.68); P ¼ 0.006 not age memory (CD45RACD27þ), effector memory
adjusted (Fig. 1c); P ¼ 0.003 age adjusted]. Telomere (CD45RACD27), and terminally differentiated
lengths were not associated with age in either ART-naive (CD45RAþCD27) cell subsets (Table 2). However,
(b ¼ 0.0054, P ¼ 0.227) or ART-treated children when central and effector memory cell subsets, the major
(b ¼ 0.0046, P ¼ 0.258) (Fig. 1d). After adjusting for cellular reservoirs for HIV [28], were considered
age, mean telomere length values tended to decrease with together, they tended to be lower in HIVþ than in
increasing HIV-RNA levels (b ¼ 0.0396; P ¼ 0.056), HEU and HUU children [21.9 (15.3–38.3)% ‘vs’ 29.8

Table 1. Demographic and clinical characteristics of HIVR, HIV-exposed-uninfected children, and HIV-unexposed-uninfected children.

HIVþ (n ¼ 71) HEU (n ¼ 65) HUU (n ¼ 56)

Age, median (IQR) years 3.11 (1.40–4.48) 1.74 (0.99–3.31) 1.85 (0.84–3.46)
Sex, n (%)
Men 39 (55%) 34 (52%) 29 (52%)
Ethnicity/race, n (%)
White 49 (69%) 47 (72.3%) 49 (87.5%)
Black 20 (28.2%) 15 (23.1%) 5 (9.0%)
Asian 2 (2.8%) 3 (4.6%) 2 (3.5%)
Exposed to ART prophylaxis, n (%) 5 (7%) 61 (93.8%) –
Exposed to ART, n (%) 41 (58%) – –
Duration of ART exposure, median (IQR) months 18 (12–36) – –
Percentage of lifetime on ART 57.5 (42.6–84.5) – –
Detectable plasmaviremia at sample collection, n (%) 54/71 (76.1%) – –
ART naive 30/30 (100%) – –
On ART 17/41 (58.5%)
Plasmaviremia at sample collection (log10 copies/ml)
ART naive 5.31 (4.90–5.62) – –
On ART 3.96 (2.70–5.27) – –

ART, antiretroviral therapy; HEU, HIV-exposed-uninfected children; HIVþ, HIV-infected children; HUU, HIV-unexposed-uninfected children;
IQR, interquartile range.
Pediatric HIV infection and aging Gianesin et al. 1367

(a) (b)
5.0 5.0
P < 0.001

Telomere length (T/S ratio)

Telomere length (T/S ratio)


4.0 4.0

3.0 3.0

2.0 2.0

1.0 1.0
HIV+ β=–0.0018, P = 0.587
HEU β=–0.0102, P = 0.008
HUU β=–0.0100, P = 0.011
0 0
HIV+ HEU HUU 0 20 40 60 80
Age (months)
(c) (d)
5.0 4.0
P = 0.006
Telomere length (T/S ratio)

Telomere length (T/S ratio)


4.0
3.0

3.0
2.0
2.0

1.0
1.0
on ART β=–0.0046, P = 0.258
0
ART-naive β=–0.0054, P = 0.227
0
ART-naive on ART 0 20 40 60 80
Age (months)
(e) (f)
20000 20000
P = 0 .018 HIV+ β=–17, P = 0.353
HEU β=–64, P = 0.009
TREC copies/105 PBMC

TREC copies/105 PBMC

HUU β=–86, P = 0.001


15000 15000

10000 10000

5000 5000

0 0
HIV+ HEU HUU 0 20 40 60 80
(g) Age (months)
(h)
15000 14000
on ART β=–8, P = 0.754
P = 0.300 ART-naive β=–11, P = 0.680
12000
TREC copies/105 PBMC

TREC copies/105 PBMC

10000
10000
8000

6000
5000
4000

2000

0 0

ART-naive on ART 0 20 40 60 80
Age (months)

Fig. 1. HIV-infected (HIVR) children have shorter telomere length (TL) and lower T-cell receptor rearrangement excision circle
(TREC) levels than HIV-exposed-uninfected (HEU) and HIV-unexposed-uninfected (HUU) children. (a) TL values not age-
adjusted in HIVþ (n ¼ 71), HEU (n ¼ 65), and HUU (n ¼ 56) children. (b) TL as function of age in HIVþ (black circles, continuous
line), in HEU (gray squares, continuous line), and HUU (white circles, dotted line) children. (c) TL values not age-adjusted in
HIVþ children, subdivided into antiretroviral therapy (ART)-naive (n ¼ 30) and on ART (n ¼ 41). (d) TL as function of age in HIVþ
children, subdivided into ART-naive (gray squares) and ART-treated (black circles). (e) TREC levels not age-adjusted in HIVþ
(n ¼ 71), HEU (n ¼ 65), and HUU (n ¼ 56) children. (f) TREC levels as function of age in HIVþ (black circles, continuous line), in
HEU (gray squares, continuous line) and HUU (white circles, dotted line) children. (g) TREC levels not age-adjusted among HIVþ
children, subdivided into ART-naive (n ¼ 30) and on ART (n ¼ 41). (h) TREC levels as function of age in HIVþ children, subdivided
into ART-naive (gray squares) and on ART (black circles). Boxes and whiskers: 25–75th and 10–90th percentiles, respectively;
central line in boxes: median. b, regression coefficient.
1368 AIDS 2016, Vol 30 No 9

Table 2. Frequencies of CD4R and CD8R T-cell subsets.

HIVþ (n ¼ 24) % median (IQR) HEU (n ¼ 21) % median (IQR) HUU (n ¼ 18) % median (IQR) Overall P value

CD3þ 64.4 (58.8–69.2) 61.8 (49.8–68.4) 60.2 (54.6–66.4) 0.590


CD4þ 39.6 (35.1–45.3) 53.4 (43.5–64.7) 52.5 (38.0–60.6) 0.001
Naive 76.8 (59.9–84.4) 70.1 (59.9–81.0) 64.7 (56.9–75.3) 0.423
Central memory 18.1 (13.2–31.1) 27.3 (17.4–32.6) 27.9 (20.7–35.1) 0.206
Effector memory 3.4 (1.2–5.8) 2.5 (1.8–6.3) 4.4 (2.9–7.8) 0.220
T. differentiated 0.5 (0.2–1.0) 0.3 (0.1–0.6) 0.4 (0.3–0.1) 0.330
RTE 63.6 (54.9–72.4) 58.3 (46.0–68.2) 54.2 (49.3–61.7) 0.181
PEC 12.4 (5.3–16.4) 11.7 (8.2–16.1) 10.4 (8.1–15.2) 0.738
Senescent 0.4 (0.1–0.7) 0.2 (0.1–0.5) 0.2 (0.1–1.3) 0.568
Activated 3.3 (2.2–5.9) 2.1 (1.3–3.5) 2.8 (1.6–3.8) 0.041
Exhausted 4.1 (3.4–6.6) 3.5 (1.9–5.2) 3.2 (2.7–5.1) 0.050
CD8þ 31.2 (25.8–39.6) 28.9 (21.2–35.0) 25.9 (22.8–29.0) 0.063
Naive 46.2 (37.6–75.1) 77.1 (55.9–84.7) 71.0 (46.7–86.1) 0.019
Central memory 11.0 (6.9–23.2) 16.3 (10.9–26.5) 12.7 (9.8–18.9) 0.278
Effector memory 7.1 (2.3–13.1) 2.1 (1.1–4.9) 4.3 (1.3–11.4) 0.033
T. differentiated 16.3 (4.5–36.4) 2.5 (1.0–8.6) 4.2 (2.1–16.2) 0.001
RTE 55.3 (41.4–71.8) 69.8 (60.4–80.1) 68.1 (59.5–79.3) 0.005
PEC 17.1 (6.5–29.2) 5.9 (3.8–15.3) 9.8 (4.6–15.2) 0.040
Senescent 25.8 (12.4–43.2) 8.5 (6.8–16.7) 9.7 (3.3–27.3) 0.004
Activated 7.0 (5.2–12.2) 4.7 (3.9–7.6) 3.4 (2.8–6.7) <0.001
Exhausted 7.1 (5.0–12.4) 3.5 (2.1–5.9) 3.7 (2.4–5.3) <0.001

HEU, HIV-exposed-uninfected children; HIVþ, HIV-infected children; HUU, HIV-unexposed-uninfected children; IQR, interquartile range; PEC,
peripheral expanded cells; RTE, recent thymic emigrant cells.

(18.6–39.4)% and 35.6 (26.2–42.6)%; P ¼ 0.085]. The HUU children (P ¼ 0.278). However, in the HIVþ
percentages of senescent cells (CD4þCD28CD57þ) group, this subset was more expanded in children with
were similar in the three groups, whereas activated detectable viremia than in those with undetectable HIV-
CD38þHLA-DRþ and exhausted PD-1þ were more RNA (P ¼ 0.027) (not shown). Both effector memory
expanded in HIVþ children than in controls (Table 2). In (CD45RACD27) and terminally differentiated cells
particular, within the HIVþ group, the percentages of (CD45RAþCD27) were more expanded in HIVþ
CD4þCD38þHLA-DRþ and CD4þPD-1þ were higher children than in control groups (Table 2). In HIVþ
in children with detectable viral load than in aviremic children, the proportion of senescent CD8þ cells was also
children (P ¼ 0.056 and P ¼ 0.037, respectively). higher than in HEU and HUU groups [25.8 (12.4–
43.2)% ‘vs.’ 8.5 (6.8–16.7)% and 9.7 (3.3–27.3)%;
The median of CD8þ cell percentages tended to be P ¼ 0.004]. This expansion was particularly observed in
higher in HIVþ children than in control groups children with detectable plasmaviremia, indicating that
(P ¼ 0.063). Notably, significant differences emerged active HIV replication stimulates the production of a
among CD8þ T-cell subsets. HIVþ children showed senescent phenotype [41.6 (18.5–45.8)% ‘vs.’ 4.4 (2.1–
lower percentage of naive cells than HEU and HUU 13.4)%; P ¼ 0.002]. In addition, the activation of CD8þ
children [46.2 (37.6–75.1)%, 77.1 (55.9–84.7)%, 71.0 cells was significantly higher in HIVþ children than in
(46.7–86.1)%; P ¼ 0.019]. In particular, HIVþ children controls [7.0 (5.2–12.2)% ‘vs.’ 4.7 (3.9–7.6)% in HEU
had a lower frequency of CD8þ recent thymic emigrant and 3.4 (2.8–6.7)% in HUU; P < 0.001] (Table 2), and
[recent thymic emigrant cells (RTE), CD45RAþCD31þ) significantly increased with HIV-RNA levels (rs ¼ 0.672;
cells (P ¼ 0.005) and a higher percentage of peripheral P < 0.001) (not shown). The percentages of CD8þPD-
expanded cells [peripheral expanded cells (PEC) 1þ cells were significantly higher in HIVþ than in HEU
CD45RAþCD31] than control groups (P ¼ 0.040), and HUU children (P ¼ 0.003 and P < 0.001, respect-
suggesting strong peripheral cell proliferation (Table 2). In ively) (Table 2). After adjustments for multiple testing, the
addition, the percentage of CD8þ RTE cells decreased CD4þ cell subset, and the CD8þ terminally differen-
with age in HEU and HUU but not in HIVþ children tiated, activated, exhausted, senescent, and RTE cell
(Fig. 2a-c ). Interestingly, CD8þ RTE cells were lower in subsets remained significantly different (P < 0.05). Also,
children with detectable viral load than in aviremic the levels of proinflammatory cytokines IL-6 and TNFa
children [41.8 (22.6–64.4)% ‘vs.’ 55.9 (53.6–76.7)%; associated with the senescent phenotype [29–31] were
P ¼ 0.039] (not shown); plasma HIV-RNA tended to be higher in HIVþ than in HEU and HUU children
inversely correlated with CD8þ RTE cells (rs ¼ 0.363, (Supplementary Table 2, http://links.lww.com/QAD/
P ¼ 0.080) and positively correlated with CD8þ PEC A905), thus supporting the accelerated immune senes-
(rs ¼ 0.357, P ¼ 0.085) (Fig. 2d). cence in HIVþ children. PD-1 expression in viremic
subjects was also significantly higher than in those with
Among memory cell subsets, the frequency of CD8þ undetectable plasmaviremia (P ¼ 0.002) and was corre-
central memory did not differ among HIVþ, HEU, and lated with HIV-RNA levels (rs ¼ 0.471, P ¼ 0.021) and
Pediatric HIV infection and aging Gianesin et al. 1369

(a) (b)
100 100

80 80
% CD8+ RTE cells

% CD8+ RTE cells


60 60

40 40

20 20
rs = −0.523 rs = −0.486
P = 0.025 P = 0.026
0 0
0 10 20 30 40 50 60 70 0 10 20 30 40 50 60 70
Age (months) Age (months)
(c) (d)
100 100 100
CD8+ RTE CD8+ PEC
rs = −0.363, P = 0.080 rs = 0.357, P = 0.085
80 80 80

% CD8+ PEC cells


% CD8+ RTE cells
% CD8+ RTE cells

60 60 60

40 40 40

20 20 20
rs = −0.242
P = 0.252
0 0 0
0 10 20 30 40 50 60 70 1 2 3 4 5 6
log10 HIV-RNA (copies/mL)
Age (months)

Fig. 2. Relationship between recent thymic emigrant (RTE) CD8R cells with age and HIV plasmaviremia. Percentage of CD8þ
RTE cells as function of age in (a) HUU, (b) HEU, and (c) HIVþ children. (d) Levels of HIV plasmaviremia in relation to percentages
of CD8þ RTE (black circles, continuous line) and CD8þ peripheral expanded cells (PEC) (white circles, dotted line) among HIVþ
children. rs, Spearman’s r correlation coefficient.

CD8þCD38þHLA-DRþ cells (rs ¼ 0.528, P ¼ 0.009) Discussion


(not shown).
This is the first study describing biological aging and
Telomere length values were inversely correlated with immune senescence in HIV-infected children compared
percentages of senescent (Fig. 3a), activated (Fig. 3d) and with HIV-exposed-uninfected and unexposed-unin-
exhausted CD8þ cells (Fig. 3g) in HIVþ, but not in HEU fected children, all aged 0–5 years. Overall, the results
(Fig. 3b, e, h) or HUU children (Fig. 3c, f, i). demonstrate that HIV-infected children exhibit prema-
ture biological aging with accelerated immune senes-
cence which affects the CD8þ T-cell subset in particular.
HIV-infected children have increased gut
microbial translocation In contrast to the study of Côté et al. [21], which found no
Median levels of sCD14 were significantly higher in difference in telomere length between HIVþ children
HIVþ than in HEU and HUU children [2699 (2333– and controls, in our study telomere length was
2826) ‘vs.’ 2138 (1954–2427) and 2065 (1850–2480) significantly shorter in HIVþ than in HEU and HUU
ng/ml; P < 0.001] (Supplementary Figure 2A, http:// children. This discordant result may be because of the
links.lww.com/QAD/A903). In HIVþ children, sCD14 different ages of the two cohorts: the children enrolled in
levels positively correlated with HIV-RNA levels our study were younger (aged 0–5 years, median 3.1)
(rs ¼ 0.585, P ¼ 0.007), with percentages of CD8þHLA- than those of Côté et al. (aged 0–19 years, median 13.3).
DRþCD38þ (rs ¼ 0.418, P ¼ 0.065), and CD8þPD-1þ As telomere shortening in peripheral blood cells is very
(rs ¼ 0.645, P ¼ 0.002), but not with percentages of rapid during the first years of life [32], the difference
CD8þCD28CD57þ (rs ¼ 0.172, P ¼ 0.487) or telomere between HIV-infected children and controls may have
length (rs ¼ 0.386, P ¼ 0.124) (Supplementary Figure emerged more clearly in our cohort. The two cohorts also
2b-f, http://links.lww.com/QAD/A903). differed in duration of ART exposure. The longer
1370 AIDS 2016, Vol 30 No 9

(a) (b) (c)


HIV+ HEU HUU
5.0 5.0 5.0
Telomere length (T/S ratio)

Telomere length (T/S ratio)

Telomere length (T/S ratio)


rs = −0.458 rs = −0.036 rs = −0.186
4.0 P = 0.054 4.0 P = 0.882 4.0 P = 0.498

3.0 3.0 3.0

2.0 2.0 2.0

1.0 1.0 1.0

0 0 0
0 10 20 30 40 50 60 0 10 20 30 40 50 0 10 20 30 40
(d) % senescent CD8+ cells (e) % senescent CD8+ cells (f) % senescent CD8+ cells

5.0
Telomere length (T/S ratio)

5.0 5.0

Telomere length (T/S ratio)

Telomere length (T/S ratio)


rs = −0.515 rs = −0.026 rs = -0.110
4.0 P = 0.019 4.0 P = 0.911 4.0 P = 0.656

3.0 3.0 3.0

2.0 2.0 2.0

1.0 1.0 1.0

0 0 0
0 5 10 15 20 25 30 0 5 10 15 20 0 5 10 15
% activated CD8+ cells % activated CD8+ cells % activated CD8+ cells
(g) (h) (i)
5.0 5.0 5.0
Telomere length (T/S ratio)

Telomere length (T/S ratio)

Telomere length (T/S ratio)


rs = −0.637 rs = −0.112 rs = −0.018
4.0 P = 0.002 4.0 P = 0.641 4.0 P = 0.934

3.0 3.0 3.0

2.0 2.0 2.0

1.0 1.0 1.0

0 0 0
0 5 10 15 20 25 30 0 5 10 15 0 5 4 6 8 10
% exhausted CD8+ cells % exhausted CD8+ cells % exhausted CD8+ cells

Fig. 3. Relationship between telomere length (TL) and T-cell immunophenotypic profiles. TL values in relation to percentages of
senescent (CD8þCD28CD57þ) cells in (a) HIVþ, (b) HEU, and (c) HUU children, activated (CD8þCD38þHLA-DRþ) cells in (d)
HIVþ, (e) HEU, and (f) HUU children, and exhausted (CD8þPD-1þ) cells in (g) HIVþ, (h) HEU, and (i) HUU children. HIVþ, HIV-
infected; HEU, HIV-exposed uninfected; HUU, HIV-unexposed uninfected; rs, Spearman’s r correlation coefficient.

exposure to ART of children in the above study (median significantly shorter telomeres than the latter. All these
85 months of ART exposure) may explain the loss of findings indicate that HIV infection per se, rather than
association found in our study population, consisting of exposure to therapy, influences the aging process. Also,
ART-naı̈ve children or ones recently on ART (median 18 the finding that over 90% of HEU children had been
months of ART exposure). perinatally exposed to ARTand that their telomere length
did not differ from those of HUU children indicates that
Nucleoside reverse transcriptase inhibitors are known relatively short-term ARTexposure does not significantly
inhibitors of telomerase reverse transcriptase, and have influence telomere length, matching the results of a
been reported to be associated with telomere shortening recent report [36]. It is possible that longer exposure to
[11,12]. The inverse association between telomere length ART in HIV-infected children exacerbates HIV-driven
and ART-exposure (median time 123 months) has telomere shortening.
recently been demonstrated in a cohort of adults, but
the small sample size and lack of optimal control of ART- The mechanism behind telomere shortening in HIV-
naive patients preclude definite conclusions [33]. Two infected individuals is unknown. Short telomeres may be
other studies of HIV-infected adults [34,35] confirm the because of excessive cellular replication after chronic
association of telomere length shortening with HIV immune activation, but the virus itself may play an active
status, but not with relatively short ART exposure role. Telomerase activity is in fact severely impaired in
(median times 58 and 48 months, respectively). uninfected CD34þ hematopoietic progenitor cells iso-
lated from HIV-infected patients [37]. HIV infection and
In our group of HIV-infected children, the ART-naive HIV-Tat protein also downmodulate telomerase expres-
ones, even though younger than those on ART, had sion and activity in lymphoblastoid cells [38] and
Pediatric HIV infection and aging Gianesin et al. 1371

peripheral blood lymphocytes [39–41]. Although the The evidence that immune senescence is more serious in
CD4þ T cell is the target of HIV infection, we found children with detectable HIV viremia focuses attention
that CD8þ T-cell compartment was largely impaired in on the need for early and long-standing control of HIV
the HIV-infected children. They had a lower frequency replication and chronic immune activation. In particular,
of CD8þ naive cells than controls and a decline in this this aspect must be considered when a treatment
cell subset did not correlate with age, as occurs in HIV- interruption has been planned [48]. As previously
uninfected children. In particular, the decreased demonstrated in a pediatric cohort, viremia does impair
percentages of RTE cells with increased percentages the immune reconstitution of memory and effector
of PEC together with increasing levels of HIV CD4þ T-cell subsets [49], and also contributes to the
plasmaviremia indicate that HIV induces peripheral expansion of T-regulatory cells [50] which may influence
proliferation of CD8þ cells and their differentiation the specific HIV immune response, allowing the virus to
into effector cells, which play a central role in replicate and increase immune activation.
immunity against pathogens [42]. As already described
in adults and older children [22,43,44], the decrease in In conclusion, HIV-infected children exhibit premature
naive cell subset is associated with skewed maturation of biological aging with accelerated immune senescence,
CD8þ cells toward an effector phenotype which, which particularly affects the CD8þ T-cell subset. The
without adequate replenishment of new CD8þ naive shorter telomere length and higher percentage of
cells, induces accumulation of cells with a senescent senescent cells in HIVþ children, compared with
phenotype. A major driver of the cellular senescent children on ART or HEU, suggest that HIV infection
phenotype is telomere shortening [29]. Our data per se, rather than exposure to ART for prophylaxis or
indicate that HIV-infected children accumulate treatment, influences the aging process. These data
CD8þCD38þ and CD8þPD-1þ cells together with a support the importance of maintaining undetectable viral
higher percentage of senescent CD8þ cells. The finding load to avoid premature immune senescence and
that activated and exhausted CD8þ cells are inversely dysfunction of CD8þ cells, compromising their tumor
correlated with telomere length supports the idea that immune surveillance function and increasing the risk of
persistent immune activation and cellular exhaustion age-related diseases, including malignancies.
are closely linked to accelerated biological aging and
immune senescence. Chronic viral coinfections may
induce immune activation and accelerate immune
senescence [35,45,46]. In particular, it has been shown Acknowledgements
that cytomegalovirus (CMV) leads to significant
changes in the CD8þ repertoire [45,46]. Unfortunately, We are grateful to Camino Estella and Francesco
clinical data on CMV serology were available only for Carmona for their skillful technical assistance in the
HIVþ and a subgroup of HEU children: 40 of 71 Hospital Sant Joan de Deu, Barcelona, and in the Unit of
(56.3%) HIVþ and nine of 33 (27.3%) HEU children Viral Oncology and AIDS Reference Center, Depart-
were CMV-positive. The CMVþ and CMV sub- ment of Surgery, Oncology and Gastroenterology.
groups of HIV-infected children did not significantly This work was supported by the Paediatric European
differ as regards telomere length and markers of Network for Treatment of AIDS (PENTA) Foundation,
immunological profile. Larger studies are needed to and from the European Union Seventh Framework
better understand the contribution of coinfections in Programme (FP7/2007–2013) under grant agreement
the aging process of HIV-infected children. n8 260694.

The finding that sCD14 levels were correlated with Authors’ contributions: K.G. designed and performed the
percentages of activated/exhausted CD8þ cells and the experiments, undertook clinical data collection, data
lack of correlation between sCD14 and senescent CD8þ analysis and interpretation, wrote the first draft, carried
cells and/or telomere length suggest that the gut damage out critical revision and provided intellectual input to
and gastrointestinal lymphocyte depletion which occurs further drafts. A.N.J. and C.F. provided clinical samples
during HIV infection do not directly cause immune and clinical data, carried out critical revision and provided
senescence, but they do contribute to accelerated aging intellectual input to further drafts. O.R., E.M., and M.C.
and senescence through immune system activation. provided clinical samples and clinical data. P.D.B.
Overall, our data support a scenario in which viremia provided statistical expertise and data analysis. Z.M.,
leads to high turnover with continual loss and output of M.R.P., and R.F. performed the experiments. C.G.
naı̈ve cells, which rapidly differentiate and exhaust their carried out critical revision and provided intellectual
effector function, resulting in an accumulation of input to further drafts. A.D.R. conceived and designed
senescent cells with short telomeres. These data are the study, carried out data analysis and interpretation,
consistent with a previous report [47] showing that high undertook critical revision and provided intellectual
percentage of CD8þCD28 correlates with shorter input to further drafts. All the authors approved the final
telomeres. version of this article.
1372 AIDS 2016, Vol 30 No 9

Conflicts of interest 17. Freguja R, Gianesin K, Zanchetta M, De Rossi A. Cross-talk


between virus and host innate immunity in pediatric HIV-1
There are no conflicts of interest. infection and disease progression. New Microbiol 2012;
35:249–257.
Partial presentation at: 22nd Conference on Retroviruses and 18. Gianesin K, Freguja R, Carmona F, Zanchetta M, Del Bianco P,
Malacrida S, et al. The role of genetic variants of stromal cell-
Opportunistic Infections, Seattle, Washington, USA, 23–26 derived factor 1 in pediatric HIV-1 infection and disease
February 2015. progression. PLoS One 2012; 7:e44460.
19. Prendergast AJ, Klenerman P, Goulder PJR. The impact of
differential antiviral immunity in children and adults. Nat
Rev Immunol 2012; 12:636–648.
20. Mansoor N, Abel B, Scriba TJ, Hughes J, de Kock M, Tameris M,
References et al. Significantly skewed memory CD8R T cell subsets in
HIV-1 infected infants during the first year of life. Clin Immunol
1. Gibb DM, Duong T, Tookey PA, Sharland M, Tudor-Williams G, 2009; 130:280–289.
Novelli V, et al. National Study of HIV in Pregnancy and 21. Côté HC, Soudeyns H, Thorne A, Alimenti A, Lamarre V, Maan
Childhood Collaborative HIV Paediatric Study. Decline in EJ, et al., the CIHR Emerging Team in HIV therapy, aging
mortality, AIDS, and hospital admissions in perinatally HIV- (CARMA). Leukocyte telomere length in HIV-infected and
1 infected children in the United Kingdom and Ireland. BMJ HIV-exposed uninfected children: shorter telomeres with un-
2003; 327:1019. controlled HIV viremia. PLoS One 2012; 7:e39266.
2. Judd A, Doerholt K, Tookey PA, Sharland M, Riordan A, Menson 22. Dı́az L, Méndez-Lagares G, Correa-Rocha R, Pacheco YM,
E, et al., Collaborative HIV Paediatric Study (CHIPS); National Ferrando-Martı́nez S, Ruiz-Mateos E, et al. Detectable viral
Study of HIV in Pregnancy and Childhood (NSHPC). Morbidity, load aggravates immunosenescence features of CD8 T-cell
mortality, and response to treatment by children in the United subsets in vertically HIV-infected children. J Acquir Immune
Kingdom and Ireland with perinatally acquired HIV infection Defic Syndr 2012; 60:447–454.
during 1996-2006: planning for teenage and adult care. Clin 23. Méndez-Lagares G, Dı́az L, Correa-Rocha R, León Leal JA,
Infect Dis 2007; 45:918–924. Ferrando-Martı́nez S, Ruiz-Mateos E, et al. Specific patterns
3. Deeks SG, Lewin RS, Havlir DV. The end of AIDS: HIV infection of CD4-associated immunosenescence in vertically HIV-
as a chronic disease. Lancet 2013; 382:1525–1533. infected subjects. Clin Microbiol Infect 2013; 19:558–565.
4. Powles T, Robinson D, Stebbing J, Shamash J, Nelson M, 24. Cawthon RM. Telomere length measurement by a novel mono-
Gazzard B, et al. Highly active antiretroviral therapy and the chrome multiplex quantitative PCR method. Nucleic Acids Res
incidence of non-AIDS-defining cancers in people with HIV 2009; 37:e21.
infection. JCO 2008; 27:884–890. 25. Rampazzo E, Bertorelle R, Serra L, Terrin L, Candiotto C,
5. Crum-Cianflone N, Hullsiek KH, Marconi V, Weintrob A, Ga- Pucciarelli S, et al. Relationship between telomere shortening,
nesan A, Barthel RV, et al. Trends in the incidence of cancers genetic instability, and site of tumour origin in colorectal
among HIV-infected persons and the impact of antiretroviral cancers. Br J Cancer 2010; 102:1300–1305.
therapy: a 20-year cohort study. AIDS 2009; 23:41–50. 26. Ruijter JM, Ramakers C, Hoogars WMH, Karlen Y, Bakker O,
6. Simard EP, Pfeiffer RM, Engels EA. Cumulative incidence of van den Hoff MJB, et al. Amplification efficiency: linking base-
cancer among people with AIDS in the United States. Cancer line and bias in the analysis of quantitative PCR data. Nucleic
2011; 117:1089–1096. Acids Res 2009; 37:e45.
7. Guaraldi G, Orlando G, Zona S, Menozzi M, Carli F, Garlassi E, 27. Ometto L, De Forni D, Patiri F, Trouplin V, Mammano F,
et al. Premature age-related comorbidities among HIV-infected Giacomet V, et al. Immune reconstitution in HIV-1-infected
persons compared with the general population. Clin Infect Dis children on antiretroviral therapy: role of thymic output and
2011; 53:1120–1126. viral fitness. AIDS 2002; 16:839–849.
8. Chiappini E, Berti E, Gianesin K, Petrara MR, Galli L, Giaquinto 28. Chomont N, El-Far M, Ancuta P, Trautmann L, Procopio FA,
C, et al. Pediatric human immunodeficiency virus infection and Yassine-Diab B, et al. HIV reservoir size and persistence are
cancer in the highly active antiretroviral treatment (HAART) driven by T cell survival and homeostatic proliferation. Nat
era. Cancer Lett 2014; 347:38–45. Med 2009; 15:893–900.
9. Effros RB, Fletcher CV, Gebo K, Halter JB, Hazzard WR, Horne 29. Campisi J, d’Adda di Fagagna F. Cellular senescence: when bad
FM, et al. Aging and infectious diseases: workshop on HIV things happen to good cells. Nat Rev Mol Cell Biol 2007; 8:729–
infection and aging: what is known and future research direc- 740.
tions. Clin Infect Dis 2008; 47:542–553. 30. Kuilman T, Peeper DS. Senescence-messaging secretome: SMS-
10. Desai S, Landay A. Early immune senescence in HIV disease. ing cellular stress. Nat Rev Cancer 2009; 9:81–94.
Curr HIV/AIDS Rep 2010; 7:4–10. 31. Young AR, Narita M. SASP reflects senescence. EMBO Rep
11. Liu X, Takahashi H, Harada Y, Ogawara T, Ogimura Y, Mizush- 2009; 10:228–230.
ina Y, et al. 3’-Azido-2’,3’-dideoxynucleoside 5’-triphosphates 32. Zeichner SL, Palumbo P, Feng Y, Xiao X, Gee D, Sleasman J,
inhibit telomerase activity in vitro, and the corresponding et al. Rapid telomere shortening in children. Blood 1999;
nucleosides cause telomere shortening in human HL60 cells. 93:2824–2830.
Nucleic Acids Res 2007; 35:7140–7149. 33. Leeansyah E, Cameron PU, Solomon A, Tennakoon S, Velayud-
12. Hukezalie KR, Thumati NR, Côté HC, Wong JM. In vitro and ex ham P, Gouillou M, et al. Inhibition of telomerase activity by
vivo inhibition of human telomerase by anti-HIV nucleoside human immunodeficiency virus (HIV) nucleos(t)ide reverse
reverse transcriptase inhibitors (NRTIs) but not by non-NRTIs. transcriptase inhibitors: a potential factor contributing to
PLoS One 2012; 7:e47505. HIV-associated accelerated aging. J Infect Dis 2013;
13. De Rossi A, Masiero S, Giaquinto C, Ruga E, Comar M, Giacca 207:1157–1165.
M, et al. Dynamics of viral replication in infants with vertically 34. Pathai S, Lawn SD, Gilbert CE, McGuinness D, McGlynn L,
acquired human immunodeficiency virus type 1 infection. Weiss HA, et al. Accelerated biological ageing in HIV-infected
J Clin Invest 1996; 97:323–330. individuals in South Africa: a case-control study. AIDS 2013;
14. McIntosh K, Shevitz A, Zaknun D, Kornegay J, Chatis P, Karthas 27:2375–2384.
N, et al. Age- and time-related changes in extracellular viral 35. Zanet DL, Thorne A, Singer J, Maan EJ, Sattha B, Le Campion A,
load in children vertically infected by human immuno- et al. Association between short leukocyte telomere length and
deficiency virus. Pediatr Infect Dis J 1996; 15:1087–1091. HIV infection in a cohort study: no evidence of a relationship
15. Henrard DR, Phillips JF, Muenz LR, Blattner WA, Wiesner D, with antiretroviral therapy. Clin Infect Dis 2014; 58:1322–
Eyster ME, et al. Natural history of HIV-1 cell-free viremia. 1332.
JAMA 1995; 274:554–558. 36. Imam T, Jitratkosol MHJ, Soudeyns H, Sattha B, Gadawski I,
16. Ricci E, Malacrida S, Zanchetta M, Montagna M, Giaquinto C, Maan E, et al. Leukocyte telomere length in HIV-infected
De Rossi A. Role of beta-defensin-1 polymorphisms in mother- pregnant women treated with antiretroviral drugs during preg-
to-child transmission of HIV-1. J Acquir Immune Defic Syndr nancy and their uninfected infants. J Acquir Immune Defic
2009; 51:13–19. Syndr 2012; 60:495–502.
Pediatric HIV infection and aging Gianesin et al. 1373

37. Vignoli M, Stecca B, Furlini G, Re MC, Mantovani V, Zauli G, 44. Montesano C, Anselmi A, Palma P, Bernardi S, Cicconi R, Mattei
et al. Impaired telomerase activity in uninfected haematopoie- M, et al. HIV replication leads to skewed maturation of
tic progenitors in HIV-1-infected patients. AIDS 1998; 12:999– CD8-positive T-cell responses in infected children. New Micro-
1005. biol 2010; 33:303–309.
38. Reynoso R, Minces L, Salomon H, Quarleri J. HIV-1 infection 45. Pawelec G, Derhovanessian E. Role of CMV in immune senes-
downregulates nuclear telomerase activity on lymphoblastoic cence. Virus Res 2011; 157:175–179.
cells without affecting the enzymatic components at the tran- 46. Solana R, Tarazona R, Aiello AE, Akbar AN, Appay V, Beswick
scriptional level. AIDS Res Hum Retrov 2006; 22:425–429. M, et al. CMV and Immunosenescence: from basics to clinics.
39. Ballon G, Ometto L, Righetti E, Cattelan AM, Masiero S, Immun Ageing 2012; 9:23.
Zanchetta, et al. Human immunodeficiency virus type 1 mod- 47. Lin J, Epel E, Cheon J, Kroenke C, Sinclair E, Bigos M,
ulates telomerase activity in peripheral blood lymphocytes. et al. Analyses and comparisons of telomerase activity and
J Infect Dis 2001; 183:417–424. telomere length in human T and B cells: insights for epide-
40. Franzese O, Adamo R, Pollicita M, Comandini A, Laudisi A, miology of telomere maintenance. J Immunol Methods 2010;
Perno CF, et al. Telomerase activity, hTERT expression, and 352:71–80.
phosphorylation are downregulated in CD4(R) T lymphocytes
48. Klein N, Sefe D, Mosconi I, Zanchetta M, Castro H, Jacobsen M,
infected with human immunodeficiency virus type 1 (HIV-1). et al. Paediatric European Network for Treatment of AIDS 11
J Med Virol 2007; 79:639–646. Trial Team. The immunological and virological consequences
41. Comandini A, Naro C, Adamo R, Akbar AN, Lanna A,
of planned treatment interruptions in children with HIV in-
Bonmassar E, et al. Molecular mechanisms involved in HIV- fection. PLoS One 2013; 8:e76582.
1-Tat mediated inhibition of telomerase activity in human CD4
(R) T lymphocytes. Mol Immunol 2013; 54:181–192. 49. Anselmi A, Vendrame D, Rampon O, Giaquinto C, Zanchetta
42. Northfield JW, Loo CP, Barbour JD, Spotts G, Hecht FM, M, De Rossi A. Immune reconstitution in human immunode-
Klenerman P, et al. Human immunodeficiency virus type 1 ficiency virus type 1-infected children with different virologi-
(HIV-1)-specific CD8R T(EMRA) cells in early infection are cal responses to antiretroviral therapy. Clin Exp Immunol 2007;
linked to control of HIV-1 viremia and predict the subsequent 150:442–450.
viral load set point. J Virol 2007; 81:5759–5765. 50. Freguja R, Gianesin K, Mosconi I, Zanchetta M, Carmona F,
43. Brenchley JM, Karandikar NJ, Betts MR, Ambrozak DR, Hill BJ, Rampon O, et al. Regulatory T cells and chronic immune
Crotty LE, et al. Expression of CD57 defines replicative senes- activation in human immunodeficiency virus 1 (HIV-1)-
cence and antigen-induced apoptotic death of CD8R T cells. infected children. Clin Exp Immunol 2011; 164:373–
Blood 2003; 101:2711–2720. 380.

Anda mungkin juga menyukai